Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(24)2021 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-34948196

RESUMO

Although guanethidine (GUA) was used in the past as a drug to suppress hyperactivity of the sympathetic nerve fibers, there are no available data concerning the possible action of this substance on the sensory component of the peripheral nervous system supplying the urinary bladder. Thus, the present study was aimed at disclosing the influence of intravesically instilled GUA on the distribution, relative frequency, and chemical coding of dorsal root ganglion neurons associated with the porcine urinary bladder. The investigated sensory neurons were visualized with a retrograde tracing method using Fast Blue (FB), while their chemical profile was disclosed with single-labeling immunohistochemistry using antibodies against substance P (SP), calcitonin gene-related peptide (CGRP), pituitary adenylate cyclase activating polypeptide (PACAP), galanin (GAL), neuronal nitric oxide synthase (nNOS), somatostatin (SOM), and calbindin (CB). After GUA treatment, a slight decrease in the number of FB+ neurons containing SP was observed when compared with untreated animals (34.6 ± 6.5% vs. 45.6 ± 1.3%), while the number of retrogradely traced cells immunolabeled for GAL, nNOS, and CB distinctly increased (12.3 ± 1.0% vs. 7.4 ± 0.6%, 11.9 ± 0.6% vs. 5.4 ± 0.5% and 8.6 ± 0.5% vs. 2.7 ± 0.4%, respectively). However, administration of GUA did not change the number of FB+ neurons containing CGRP, PACAP, or SOM. The present study provides evidence that GUA significantly modifies the sensory innervation of the porcine urinary bladder wall and thus may be considered a potential tool for studying the plasticity of this subdivision of the bladder innervation.


Assuntos
Gânglios Espinais/metabolismo , Guanetidina/farmacologia , Bexiga Urinária/inervação , Antagonistas Adrenérgicos/farmacologia , Neurônios Adrenérgicos/efeitos dos fármacos , Neurônios Adrenérgicos/metabolismo , Animais , Calbindinas/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Feminino , Galanina/metabolismo , Gânglios Espinais/efeitos dos fármacos , Guanetidina/metabolismo , Neurotoxinas/farmacologia , Óxido Nítrico Sintase/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Células Receptoras Sensoriais/metabolismo , Somatostatina/metabolismo , Substância P/metabolismo , Suínos , Bexiga Urinária/efeitos dos fármacos
2.
PLoS One ; 13(9): e0203573, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30199552

RESUMO

Endothelium is the main source of catecholamine release in the electrical-field stimulation (EFS)-induced aortic contractions of the non- venomous snake Panterophis guttatus. However, adrenergic vasomotor control in venomous snakes such as Crotalus durissus terrificus and Bothrops jararaca has not yet been investigated. Crotalus and Bothrops aortic rings were mounted in an organ bath system. EFS-induced aortae contractions were performed in the presence and absence of guanethidine (30 µM), phentolamine (10 µM) or tetrodotoxin (1 µM). Frequency-induced contractions were also performed in aortae with endothelium removed. Immunohistochemical localization of both tyrosine hydroxylase (TH) and S-100 protein in snake aortic rings and brains, as well as in human tissue (paraganglioma tumour) were carried out. EFS (4 to 16 Hz) induced frequency-dependent aortic contractions in both Crotalus and Bothrops. The EFS-induced contractions were significantly reduced in the presence of either guanethidine or phentolamine in both snakes (p<0.05), whereas tetrodotoxin had no effect in either. Removal of the endothelium abolished the EFS-induced contractions in both snakes aortae (p<0.05). Immunohistochemistry revealed TH localization in endothelium of both snake aortae and human vessels. Nerve fibers were not observed in either snake aortae. In contrast, both TH and S100 protein were observed in snake brains and human tissue. Vascular endothelium is the main source of catecholamine release in EFS-induced contractions in Crotalus and Bothrops aortae. Human endothelial cells also expressed TH, indicating that endothelium- derived catecholamines possibly occur in mammalian vessels.


Assuntos
Aorta/efeitos dos fármacos , Bothrops/metabolismo , Catecolaminas/metabolismo , Crotalus/metabolismo , Estimulação Elétrica , Animais , Catecolaminas/farmacologia , Endotélio Vascular/efeitos dos fármacos , Guanetidina/metabolismo , Guanetidina/farmacologia , Técnicas In Vitro , Fentolamina/metabolismo , Fentolamina/farmacologia , Proteínas S100/metabolismo , Tetrodotoxina/metabolismo , Tetrodotoxina/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo
3.
Bone ; 41(2): 206-15, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17543595

RESUMO

The involvement of the sympathetic nervous system (SNS) in the modulation of bone adaptation to its load-bearing demand remains controversial. This study tested the involvement of SNS in the adaptive response of trabecular and cortical bone to either external loading or disuse. External loading consisted of cyclic strain (40 cycles, peak 1500 microstrain) applied for 7 min, 3 days/week, while disuse was induced by unilateral sciatic neurectomy (SN). C57Bl/J6 mice, female, 9 weeks old, were subjected to loading or disuse for 2 weeks. Half of the loaded and SN mice were injected with the beta-adrenergic antagonist, propranolol (PRO, 20 mug/g) 1 week before the start of loading or disuse and during all the duration of the experiment. MicroCT analysis of the tibiae showed that the applied load induced significant changes on both trabecular architecture and cortical geometry compared to the contralateral controls, indicating increased bone mass. In contrast, disuse markedly reduced trabecular and cortical indexes. However, these adaptive responses were not altered by PRO treatment. We further tested whether the lack of protective effect of PRO against disuse-induced bone loss was due to the very short duration of treatment by blocking SNS signaling for 8 weeks with either PRO (0.5 mg/ml in drinking water) or guanethidine sulfate (GS, 40 mug/g, injected). At the end of fourth week of treatment, mice underwent SN surgery so that disuse was induced for the remaining 4 weeks. Again, neither PRO nor GS treatments altered the disuse-induced bone loss in the neurectomized tibia. In addition, blockade of SNS signaling for either 3 or 8 weeks did not affect the basal trabecular bone architecture in control tibiae and in L4 vertebrae. This study shows that the mechano-adaptive response occurring in trabecular and cortical bone upon loading or disuse is not altered by inactivation of beta-adrenergic signaling. Furthermore, sympathectomy had no effect on trabecular bone at different skeletal sites. This suggests that the osteo-regulatory action of beta-adrenergic signaling is not involved in the bone mechano-adaptive response and must therefore affect other bone regulatory pathways.


Assuntos
Adaptação Fisiológica , Antagonistas Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/fisiologia , Suporte de Carga , Adrenérgicos/metabolismo , Animais , Peso Corporal , Feminino , Guanetidina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Propranolol/metabolismo , Coluna Vertebral/anatomia & histologia , Coluna Vertebral/fisiologia , Estresse Mecânico , Tíbia/anatomia & histologia , Tíbia/fisiologia , Tomografia Computadorizada por Raios X
4.
J Biol Chem ; 274(8): 4876-82, 1999 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-9988729

RESUMO

The question of which is the active form of dopamine for the neuronal dopamine transporter is addressed in HEK-293 cells expressing the human dopamine transporter. The Km value for [3H]dopamine uptake fell sharply when the pH was increased from 6.0 to 7.4 and then changed less between pH 7.4 and 8.2. The KI for dopamine in inhibiting the cocaine analog [3H]2beta-carbomethoxy-3beta-(4-fluorophenyl)tropane binding displayed an identical pH dependence, suggesting that changes in uptake result from changes in dopamine recognition. Dopamine can exist in the anionic, neutral, cationic, or zwitterionic form, and the contribution of each form was calculated. The contribution of the anion is extremely low (

Assuntos
Proteínas de Transporte/metabolismo , Dopamina/metabolismo , Glicoproteínas de Membrana , Proteínas de Membrana Transportadoras , Proteínas do Tecido Nervoso , Sítios de Ligação , Compostos de Bretílio/metabolismo , Linhagem Celular , Cocaína/análogos & derivados , Cocaína/metabolismo , Dopamina/química , Proteínas da Membrana Plasmática de Transporte de Dopamina , Inibidores da Captação de Dopamina/metabolismo , Guanetidina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Neurônios/metabolismo
5.
J Pharmacol Exp Ther ; 253(3): 913-20, 1990 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1972754

RESUMO

Experiments were performed to evaluate the mechanism underlying our recent observation that reserpine but not surgical denervation up-regulates rat renal cortical beta adrenergic receptors. The specific binding of [125I]iodocyanopindolol was used to quantitate the beta adrenoceptors. Chronic high-dose guanethidine, which decreased renal tissue and circulating catecholamines to the same extent as reserpine, failed to up-regulate renal beta adrenoceptors, which indicates that the effect of reserpine was not due to changes in the ambient catecholamine concentration. Isoproterenol-stimulated renin secretion, a measure of postsynaptic receptor function, was increased by reserpine but not denervation, which indicates that the failure to observe beta adrenoceptor up-regulation by radioligand binding studies after denervation was not an anomaly caused by loss of presynaptic receptors masking postsynaptic supersensitivity. Reserpine was effective even in denervated kidneys. Up-regulation of renal beta adrenoceptors with reserpine occurred even after destruction of peripheral sympathetic nervous system by a combination of adrenal demedullation and high-dose guanethidine administration. A lower daily dose of reserpine (0.3 mg/kg instead of 0.5 mg/kg), which caused no weight loss, was effective in producing beta adrenoceptor up-regulation. Antagonism of reserpine depletion of nerve terminal norepinephrine by tranylcypromine, a monoamine oxidase inhibitor, did not nullify renal beta adrenoceptor up-regulation. Overall, our results indicate that reserpine up-regulation of renal beta adrenergic receptors is independent of the sympathetic nervous system and possibly is a direct effect.


Assuntos
Córtex Renal/efeitos dos fármacos , Receptores Adrenérgicos beta/efeitos dos fármacos , Reserpina/farmacologia , Sistema Nervoso Simpático/efeitos dos fármacos , Antagonistas Adrenérgicos beta/farmacologia , Animais , Ligação Competitiva , Catecolaminas/sangue , Catecolaminas/fisiologia , Guanetidina/metabolismo , Guanetidina/farmacologia , Córtex Renal/metabolismo , Masculino , Pindolol/análogos & derivados , Pindolol/farmacologia , Ratos , Ratos Endogâmicos , Receptores Adrenérgicos beta/metabolismo , Sistema Nervoso Simpático/metabolismo , Regulação para Cima/efeitos dos fármacos
6.
J Pharm Pharmacol ; 39(12): 1052-5, 1987 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2894440

RESUMO

The capacity of human liver microsomes to N-oxidize guanethidine from 25 subjects has been assessed. Guanethidine N-oxidation was optimal at pH 8.5 and proceeded at only 16% of the maximal rate at pH 7.4. The mean rates of guanethidine N-oxidation at pH 8.5 and 7.4 were 2.46 +/- 0.89 (mean +/- s.d., n = 25) and 0.38 +/- 0.22 (mean +/- s.d., n = 22), respectively. Interindividual differences in the rate of guanethidine N-oxidation at pH 8.5 and 7.4 were 17- and 11-fold, respectively. The cytochrome P450 inhibitors, proadifen and 2,4-dichloro-6-phenylphenoxyethylamine (DPEA), at both pH 8.5 and 7.4 caused less than 20% reduction in the rate of guanethidine N-oxidation by human liver microsomes. These data indicate that guanethidine N-oxidation can be used as a measure of flavin-containing monooxygenase activity in human liver.


Assuntos
Guanetidina/metabolismo , Microssomos Hepáticos/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Microssomos Hepáticos/enzimologia , Oxirredução
7.
Brain Res ; 383(1-2): 100-9, 1986 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-3768685

RESUMO

Guanacline, a guanidinium adrenergic neuron blocking agent similar to guanethidine, was studied clinically and experimentally during the late 1960s. Like guanethidine, it has been reported to produce sympathetic neuronal destruction in rats. Unlike guanethidine, it has been reported to produce irreversible sympathetic deficits in man and to produce fluorescent lipopigment in rat sympathetic neurons. Guanacline and its derivative in which the double bond of the tetrahydropyridine ring is reduced (saturated analog of guanacline, SAG) were prepared. Several species were treated chronically with varying doses of guanethidine, guanacline or SAG; the superior cervical ganglia were examined light microscopically for neuronal destruction and for osmiophilic fluorescent lipopigment accumulation. All 3 drugs produced rapid neuronal destruction in rats accompanied by massive small-cell infiltration. In striking contrast, treatment for many weeks with doses up to 100 mg/kg/day produced no small-cell infiltration or apparent neuronal destruction in mice or guinea pigs. The neuronal destruction produced by guanacline and SAG in the rat, like that caused by guanethidine, was prevented by immunosuppression or gamma-irradiation, indicating that all 3 agents produce neuronal destruction in rats by an immune-mediated mechanism. Thus, the ability of the drug to produce sympathectomy is species specific but not drug specific. The opposite was found with respect to fluorescent lipopigment accumulation. Guanacline, but not guanethidine or SAG, produced fluorescent lipopigment in all species examined. Therefore, the double bond of the tetrahydropyridine ring plays a critical role in the production of the fluorescent lipopigment.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Gânglios Simpáticos/metabolismo , Guanidinas/farmacologia , Imunossupressores , Pigmentos Biológicos/metabolismo , Simpatectomia Química , Simpatolíticos/farmacologia , Animais , Cricetinae , Guanetidina/metabolismo , Guanidinas/metabolismo , Cobaias , Camundongos , Ratos , Ratos Endogâmicos , Especificidade da Espécie
8.
Brain Res ; 340(1): 61-9, 1985 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-3161586

RESUMO

The chronic administration of guanethidine causes an immune-mediated destruction of sympathetic neurons in rats. Destruction can be prevented by various immunosuppressive agents, including gamma-irradiation and cyclophosphamide, as well as by administration with nerve growth factor (NGF). Experiments were conducted to determine whether: (1) NGF prevented accumulation of guanethidine within sympathetic neurons; and (2) NGF caused an inhibition of immune function by either blocking proliferation of immune-competent cells or by blocking effector function even in the presence of antigen and activated immune cells. NGF did not prevent accumulation of guanethidine within sympathetic ganglia in vivo, a necessary prerequisite for neuronal destruction, nor was it inhibitory on immune function using several assay systems. NGF, purified by either conventional methods or additionally by HPLC ("ultrapure'), did not inhibit either proliferation of cloned cytotoxic T lymphocytes (CTL) to antigen (class I major histocompatibility antigens) or lysis of target cells bearing the appropriate antigens. In addition, NGF did not exhibit growth stimulating effects in this assay system (i.e. it could not substitute for T cell growth factor). NGF also did not cause an inhibition of either murine or rat allogeneic mixed lymphocyte responses measured by lysis of appropriate target cells or proliferation, respectively. Finally, NGF did not inhibit, but rather appeared to stimulate the antibody response to sheep red blood cells generated in vivo in young rats. Thus NGF does not appear to prevent the immune-mediated neural destruction induced by guanethidine by acting as an immunosuppressive agent, but rather acts by some other mechanism such as preventing expression or recognition of antigen(s) on the sympathetic neuron.


Assuntos
Guanetidina , Degeneração Neural/efeitos dos fármacos , Fatores de Crescimento Neural/uso terapêutico , Neurônios/imunologia , Simpatectomia Química , Animais , Guanetidina/metabolismo , Técnicas In Vitro , Ativação Linfocitária/efeitos dos fármacos , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Neurônios/efeitos dos fármacos , Ratos , Ratos Endogâmicos , Linfócitos T Citotóxicos/imunologia
9.
Drugs ; 30(1): 22-31, 1985 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-3896742

RESUMO

Guanadrel sulphate is an orally active peripheral sympathetic inhibitor (adrenergic neuron-blocking drug). In comparative studies, guanadrel was comparable in efficacy with guanethidine or methyldopa in mild to moderately severe hypertension, although generally it caused fewer central nervous system side effects than methyldopa and less orthostatic dizziness and diarrhoea than guanethidine. However, its efficacy in patients whose blood pressure remains inadequately controlled by other drugs (except diuretics alone) has yet to be adequately demonstrated. Guanadrel has a rapid onset of action and a half-life of about 10 hours, thus dose titration can be achieved more rapidly than with guanethidine, and twice daily administration is appropriate. Generally, guanadrel has been well tolerated, withdrawal of treatment due to adverse effects seldom being necessary. Thus, guanadrel appears to be a suitable alternative to methyldopa for the treatment of mild to moderately severe hypertension not controlled adequately by diuretics alone.


Assuntos
Anti-Hipertensivos/farmacologia , Guanidinas/farmacologia , Hipertensão/tratamento farmacológico , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/efeitos adversos , Anti-Hipertensivos/metabolismo , Anti-Hipertensivos/intoxicação , Anti-Hipertensivos/uso terapêutico , Sistema Nervoso Central/efeitos dos fármacos , Interações Medicamentosas , Guanetidina/metabolismo , Guanetidina/uso terapêutico , Guanidinas/administração & dosagem , Guanidinas/efeitos adversos , Guanidinas/metabolismo , Guanidinas/intoxicação , Guanidinas/uso terapêutico , Hemodinâmica/efeitos dos fármacos , Humanos , Cinética , Metildopa/uso terapêutico , Sistema Nervoso Simpático/efeitos dos fármacos
10.
J Med Chem ; 27(2): 149-55, 1984 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-6694163

RESUMO

Fourteen 125I-labeled aralkylguanidines were synthesized and evaluated as potential imaging agents for the adrenal medullae and tumors of adrenomedullary origin. These guanidines are radiotracer analogues of guanethidine, an antihypertensive agent thought to mediate neuron blockade by uptake into adrenergic nerves. Dog adrenal medullae were used as a model to test radiotracer affinity for catecholamine storage tissue. Tissue distribution studies revealed that a number of radioiodinated guanidines showed pronounced localization in the adrenal medullae following intravenous injection, in certain cases exceeding that of either (-)-[3H]norepinephrine or [14C]guanethidine. (m-[125I]Iodobenzyl)guanidine (m-IBG, 2b) gave the best combination of high concentration and selectivity. The low adrenomedullary affinity observed with [14C]guanidine and m-[125I]iodobenzylamine demonstrates the uniqueness of the aralkylguanidine structure. Preliminary evidence suggests that 2b is a storage analogue of norepinephrine. [125I]2a is now being used clinically in imaging and radiotherapy of catecholamine tumors, such as pheochromocytoma.


Assuntos
Medula Suprarrenal/metabolismo , Radioisótopos do Iodo , Iodobenzenos/metabolismo , 3-Iodobenzilguanidina , Neoplasias das Glândulas Suprarrenais/diagnóstico por imagem , Medula Suprarrenal/diagnóstico por imagem , Animais , Grânulos Cromafim/metabolismo , Cães , Guanetidina/análogos & derivados , Guanetidina/metabolismo , Norepinefrina/metabolismo , Cintilografia , Relação Estrutura-Atividade , Distribuição Tecidual
12.
Mayo Clin Proc ; 55(6): 390-402, 1980 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-6103981

RESUMO

Antihypertensive drugs that alter the function of the adrenergic neuron make up one of the largest groups of medications used to treat elevated blood pressure. The mechanisms of action and clinical pharmacology of widely used antihypertensive drugs that interfere with adrenergic function are reviewed. Emphasis is placed on the importance of an understanding of the biochemistry of the adrenergic neuron and the subclassification of adrenoceptors for an appreciation of the mechanism of action and the side effects of these drugs.


Assuntos
Anti-Hipertensivos/farmacologia , Sistema Nervoso Simpático/efeitos dos fármacos , Antagonistas Adrenérgicos beta/farmacologia , Catecolaminas/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Clonidina/farmacologia , Guanetidina/metabolismo , Guanetidina/farmacologia , Humanos , Metildopa/farmacologia , Metoprolol/farmacologia , Nadolol , Prazosina/metabolismo , Prazosina/farmacologia , Propanolaminas/farmacologia , Propranolol/farmacologia , Receptores Adrenérgicos/fisiologia , Reserpina/farmacologia , Sistema Nervoso Simpático/metabolismo
13.
Eur J Clin Pharmacol ; 15(2): 121-5, 1979 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-374089

RESUMO

The plasma level and urinary excretion rate of guanethidine have been measured in 30 patients during oral maintenance therapy, and in 5 patients following discontinuous of therapy. A significant correlation was found between the daily average urinary excretion and the maintenance dose, although wide interindividual variation was noted among patients maintained on the same dose. A statistically significant correlation was also observed between the area under the plasma level curve during the dose interval and the oral maintenance dose. After discontinuation of chronic therapy, the half-life of 1.5 days of the initial phase of elimination was essentially in agreement with the half-life of almost 2 days determined in acute studies. In addition, a second phase of elimination with a half-life of 4 to 8 days was observed.


Assuntos
Guanetidina/metabolismo , Administração Oral , Ensaios Clínicos como Assunto , Guanetidina/administração & dosagem , Guanetidina/uso terapêutico , Humanos , Hipertensão/tratamento farmacológico , Fatores de Tempo
14.
J Cell Biol ; 79(1): 121-31, 1978 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-701368

RESUMO

Cultures of dissociated rat superior cervical ganglion neurons (SCGN) were treated with the sympatholytic agent, guanethidine. When treated within the first couple of weeks in vitro, the neurons were rapidly destroyed. The cells grew less susceptible to the toxic effects of guanethidine with age in vitro. Moreover, the apparent affinity, Km, of the transport molecule for norepinephrine (NE) and guanethidine remained essentially unchanged between 2 and 7 wk in culture, as did the maximum velocity of transport (Vmax). This is at a time when previous studies have shown these neurons to be using acetylcholine (ACh) as their neurotransmitter. Cultures which were grown without supporting cells and from which cholinergic synaptic interactions were recorded physiologically were processed for autoradiography after incubation with [3H]NE. All cell bodies and processes seen had silver grains accumulated over them. These experiments show that sympathetic neurons in vitro maintain their amine uptake system relatively unchanged, even though they use ACh as their transmitter. The implications of these findings are discussed.


Assuntos
Acetilcolina/fisiologia , Aminas/metabolismo , Guanetidina/farmacologia , Neurônios/metabolismo , Sistema Nervoso Simpático/citologia , Transporte Biológico Ativo , Células Cultivadas , Guanetidina/metabolismo , Cinética , Neurônios/efeitos dos fármacos , Norepinefrina/metabolismo , Sistema Nervoso Simpático/metabolismo
15.
Wien Klin Wochenschr ; 89(8): 260-5, 1977 Apr 15.
Artigo em Alemão | MEDLINE | ID: mdl-857432

RESUMO

A. The clinical applications of oxygen under high pressure (OHP) are limited by oxygen toxicity. Hence, an investigation was carried out in mice on the influence of drugs on the lethal effect of OHP. 1. The lethal effect of OHP is diminished by phenobarbitone, propranolol, clonidine, succinate and tris buffer. 2. The lethal effect of OHP is enhanced by methamphetamine, acetazolamide and guanethidine. 3. The lethal effect of OHP is enhanced by reserpine two hours after administration, but diminished 12 hours after administration. B. The clinical usage of OHP is often necessarily connected with drug therapy. Hence, alteration in drug effects under OHP were investigated in mice and rats. 1. The convulsion threshold of pentetrazol is reduced under OHP by 26%. 2. The duration of the hypnotic effect of hexobarbitone is reduced under OHP by 27%. 3. The analgesic effect of morphine is unchanged by OHP. Practical aspects with regard to the use of drugs during clinical use of OHP are discussed.


Assuntos
Interações Medicamentosas , Oxigenoterapia Hiperbárica , Oxigênio/toxicidade , Acetazolamida/metabolismo , Animais , Clonidina/metabolismo , Antagonismo de Drogas , Sinergismo Farmacológico , Guanetidina/metabolismo , Metanfetamina/metabolismo , Camundongos , Fenobarbital/metabolismo , Propranolol/metabolismo , Reserpina/metabolismo , Succinatos/metabolismo , Tetrazóis/metabolismo
16.
Br J Pharmacol ; 58(4): 497-504, 1976 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1000128

RESUMO

1 The binding and efflux characteristics of [14C]-guanethidine and [3H]-noradrenaline were studied in heart slices from rats which were pretreated with reserpine and nialamide. 2 Binding of both compounds occurred at extra-vesicular sites within the adrenergic neurone. After a brief period of rapid washout, the efflux of [14C]-guanethidine and [3H]-noradrenaline proceeded at a steady rate. The efflux of both compounds appeared to occur from a single intraneuronal compartment. 3 (+)-Amphetamine accelerated the efflux of [14C]-noradrenaline; this effect was inhibited by desipramine. 4 Unlabelled guanethidine and amantadine also increased the efflux of labelled compounds. Cocaine in high concentrations increased slightly the efflux of [14C]-guanethidine but not that of [3H]-noradrenaline. 5 Heart slices labelled with [3H]-noradrenaline became refractory to successive exposures to releasing agents although an appreciable amount of labelled compound was still present in in these slices. 6 It is suggested that [14C]-guanethidine and [3H]-noradrenaline are bound at a common extravesicular site within the adrenergic neurone. Binding of guanethidine to the extra-vesicular site may be relevant to its pharmacological action, i.e., the blockade of adrenergic transmission.


Assuntos
Sítios de Ligação/efeitos dos fármacos , Guanetidina/metabolismo , Miocárdio/metabolismo , Neurônios/metabolismo , Norepinefrina/metabolismo , Anfetamina/farmacologia , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Feminino , Coração/inervação , Técnicas In Vitro , Masculino , Membranas/metabolismo , Miocárdio/citologia , Ratos , Estimulação Química , Fatores de Tempo
17.
Schweiz Med Wochenschr ; 106(45): 1531-8, 1976 Nov 06.
Artigo em Alemão | MEDLINE | ID: mdl-65008

RESUMO

Cardiovascular therapy frequently requires simultaneous administration of several drugs. These drugs may interact pharmacokinetically or pharmacodynamically, and most drug interactions are therapeutically useful. The clinical importance of adverse drug interactions is often overestimated; they are almost always predictable and preventable, if their mechanisms are understood and the drug dosages appropriately modified. Coumarin anticoagulants and cardiac glycosides are involved in the most serious interactions. Diuretics, antiarrhythmic drugs, sympatholytics, sympathomimetics and monoamine oxidase blockers also interact in clinically significant fashion with other drugs.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Interações Medicamentosas , Antidepressivos/farmacologia , Transporte Biológico , Biotransformação , Dicumarol/metabolismo , Digoxina/metabolismo , Guanetidina/metabolismo , Humanos , Absorção Intestinal , Cinética , Ligação Proteica , Albumina Sérica
19.
Pharmacol Biochem Behav ; 3(2): 205-28, 1975.
Artigo em Inglês | MEDLINE | ID: mdl-1096173

RESUMO

Marked differences in extent of diffusion have been shown with the fluorescence histochemical method between guanethidine and 6-OHDA(64 mug in 2 mul) when injected acutely or chronically into the lateral hypothalamus, the substantia nigra or the amygdala of the rat brain. Cannulation damage up to 1 mm in diameter and attributed to the implantation of cannulae and placebo injection was observed. A further area of generalized damage occurred following the injection of drugs and was far greater for 6-OHDA (2 mm) than for guanethidien (0.3 mm). Guanethidine, but not 6-OHDA, caused specific damage to catecholamine-containing nerurons up to a distance of at least 3 mm and more from the cannula tip. These striking differences between the effects of intracranial injection of 6-OHDA and guanethidine are discussed in terms of the uptake and degradation of the two drugs and the anatomical features of the injection site; they are not explicable in terms of experimental conditions such as concentration, volume of injection, molecular weight or lipid solubility. The different patterns of damage would not easily be distinguished by biochemical analyses and the catecholamine specificity of 6-OHDA in studies of the central nervous system must be seriously questioned. Vascularization of chronically implanted cannula tracks and the presence of anatomical diffusion barriers are also discussed in relation to the diffusion of drugs injected intracranially.


Assuntos
Tonsila do Cerebelo/metabolismo , Guanetidina/administração & dosagem , Hidroxidopaminas/administração & dosagem , Hipotálamo/metabolismo , Degeneração Neural , Técnicas Estereotáxicas , Substância Negra/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Química Encefálica/efeitos dos fármacos , Catecolaminas/análise , Difusão , Guanetidina/metabolismo , Guanetidina/farmacologia , Histocitoquímica , Hidroxidopaminas/metabolismo , Hidroxidopaminas/farmacologia , Hipotálamo/efeitos dos fármacos , Injeções , Masculino , Microscopia Eletrônica , Microscopia de Fluorescência , Degeneração Neural/efeitos dos fármacos , Ratos , Técnicas Estereotáxicas/efeitos adversos , Substância Negra/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA